1
|
Li N, Chen S, Cai X. Harnessing molecular probes for imaging of human epidermal growth factor receptor (HER) family. Bioorg Med Chem 2024; 113:117931. [PMID: 39362074 DOI: 10.1016/j.bmc.2024.117931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/15/2024] [Accepted: 09/16/2024] [Indexed: 10/05/2024]
Abstract
The human epidermal growth factor receptor (HER) family plays a critical role in the development, migration, and invasion of various cancers. Currently, the FDA has approved numerous targeting therapies for the HER family consist of small molecule drugs, monoclonal antibodies and antibody-drug conjugates. To facilitate precision therapy using currently approved targeted agents, early detection and quantification of each HER receptor are essential for assessment, treatment, and prognostic purposes. This study provides a comprehensive review of the latest advancements in detection and quantification of HER receptors, including traditional biopsies, liquid biopsies, and non-invasive detection methods. Although traditional histological methods, such as immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH), have yielded valuable insights, advancements in real-time and non-invasive detection technologies necessitate improved methods for the dynamic evaluation of HER status. This article also reviews several emerging real-time techniques for detecting and quantifying HER status in circulating tumor cells (CTCs) extracted from blood samples, as well as in vivo assessments using positron emission tomography (PET) and single-photon emission computed tomography (SPECT) imaging. This review emphasizes the importance of continuous innovation in the application of HER receptor imaging technologies, with the goal of enhancing treatment outcomes and prognoses for cancer patients.
Collapse
Affiliation(s)
- Na Li
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China
| | - Shengxi Chen
- Biodesign Center for BioEnergetics, Arizona State University, Tempe 85287, USA.
| | - Xiaoqing Cai
- School of Pharmaceutical Sciences, Sun Yat-sen University, 132 East Outer Ring Road, Guangzhou 510006, China.
| |
Collapse
|
2
|
Takla FN, Bayoumi WA, El-Messery SM, Nasr MNA. Developing multitarget coumarin based anti-breast cancer agents: synthesis and molecular modeling study. Sci Rep 2023; 13:13370. [PMID: 37591917 PMCID: PMC10435442 DOI: 10.1038/s41598-023-40232-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/07/2023] [Indexed: 08/19/2023] Open
Abstract
A new series of 7-substituted coumarin scaffolds containing a methyl ester moiety at the C4-position were synthesized and tested for their in vitro anti-proliferative activity against MCF-7 and MDA-MB-231 breast cancer cell lines using Doxorubicin (DOX) as reference. Compounds 2 and 8 showed noticeable selectivity against MCF-7 with IC50 = 6.0 and 5.8 µM, respectively compared to DOX with IC50 = 5.6 µM. Compounds 10, 12, and 14 exhibited considerable selectivity against Estrogen Negative cells with IC50 = 2.3, 3.5, and 1.9 µM, respectively) compared to DOX with (IC50 = 7.3 µM). The most promising compounds were tested as epidermal growth factor receptor and aromatase (ARO) enzymes inhibitors using erlotinib and exemestane (EXM) as standards, respectively. Results proved that compound 8 elicited the highest inhibitory activity (94.73% of the potency of EXM), while compounds 10 and 12 displayed 97.67% and 81.92% of the potency of Erlotinib, respectively. Further investigation showed that the promising candidates 8, 10, and 12 caused cell cycle arrest at G0-G1 and S phases and induced apoptosis. The mechanistic pathway was confirmed by elevating caspases-9 and Bax/Bcl-2 ratio. A set of in silico methods was also performed including docking, bioavailability ADMET screening and QSAR study.
Collapse
Affiliation(s)
- Fiby N Takla
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Delta University for Science and Technology, International Coastal Road, Gamasa City, 35712, Egypt
| | - Waleed A Bayoumi
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Shahenda M El-Messery
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt.
| | - Magda N A Nasr
- Department of Pharmaceutical Organic Chemistry, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| |
Collapse
|
3
|
Yu G, Lyu Y, Jiang L, Wang Y, Yin Y, Zhang J, Yang M, Tang F. ELISA-like QDB method to meet the emerging need of Her2 assessment for breast cancer patients. Front Oncol 2023; 13:920698. [PMID: 36969021 PMCID: PMC10036774 DOI: 10.3389/fonc.2023.920698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 02/21/2023] [Indexed: 03/12/2023] Open
Abstract
Inherent issues of subjectivity and inconsistency have long plagued immunohistochemistry (IHC)-based Her2 assessment, leading to the repeated issuance of guidelines by the American Society of Clinical Oncology/College of American Pathologists (ASCO/CAP) for its standardization for breast cancer patients. Yet, all these efforts may prove insufficient with the advent of Trastuzumab deruxtecan (T-Dxd), a drug with the promise to expand to tumors traditionally defined as Her2 negative (Her2-). In this study, we attempted to address these issues by exploring an ELISA-like quantitative dot blot (QDB) method as an alternative to IHC. The QDB method has been used to measure multiple protein biomarkers including ER, PR, Ki67, and cyclin D1 in breast cancer specimens. Using an independent cohort (cohort 2) of breast cancer formalin-fixed paraffin-embedded (FFPE) specimens, we validated cutoffs developed in cohort 1 (Yu et al., Scientific Reports 2020 10:10502) with overall 100% specificity (95% CI: 100-100) and 97.56% sensitivity (95% CI: 92.68-100) in cohort 2 against standard practice with the dichotomized absolutely quantitated values. Using the limit of detection (LOD) of the QDB method as the putative cutoff point, tumors with no Her2 expression were identified with the number comparable to those of IHC 0. Our results support further evaluation of the QDB method as an alternative to IHC to meet the emerging need of identifying tumors with low Her2 expression (Her2-low) in daily clinical practice.
Collapse
Affiliation(s)
- Guohua Yu
- Laboratory of Molecular Pathology, Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Yan Lyu
- Yantai Quanticision Diagnostics, Inc., a Division of Quanticision Diagnostics, Inc. (US), Yantai, Shandong, China
| | - Lei Jiang
- Laboratory of Molecular Pathology, Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Yunjun Wang
- Laboratory of Molecular Pathology, Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Ying Yin
- Laboratory of Molecular Pathology, Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
| | - Jiandi Zhang
- Yantai Quanticision Diagnostics, Inc., a Division of Quanticision Diagnostics, Inc. (US), Yantai, Shandong, China
| | - Maozhou Yang
- Yantai Quanticision Diagnostics, Inc., a Division of Quanticision Diagnostics, Inc. (US), Yantai, Shandong, China
| | - Fangrong Tang
- Yantai Quanticision Diagnostics, Inc., a Division of Quanticision Diagnostics, Inc. (US), Yantai, Shandong, China
| |
Collapse
|
4
|
Yousefi H, Bahramy A, Zafari N, Delavar MR, Nguyen K, Haghi A, Kandelouei T, Vittori C, Jazireian P, Maleki S, Imani D, Moshksar A, Bitaraf A, Babashah S. Notch signaling pathway: a comprehensive prognostic and gene expression profile analysis in breast cancer. BMC Cancer 2022; 22:1282. [PMID: 36476410 PMCID: PMC9730604 DOI: 10.1186/s12885-022-10383-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022] Open
Abstract
Breast cancer is a complex disease exhibiting a great degree of heterogeneity due to different molecular subtypes. Notch signaling regulates the differentiation of breast epithelial cells during normal development and plays a crucial role in breast cancer progression through the abnormal expression of the Notch up-and down-stream effectors. To date, there are only a few patient-centered clinical studies using datasets characterizing the role of Notch signaling pathway regulators in breast cancer; thus, we investigate the role and functionality of these factors in different subtypes using publicly available databases containing records from large studies. High-throughput genomic data and clinical information extracted from TCGA were analyzed. We performed Kaplan-Meier survival and differential gene expression analyses using the HALLMARK_NOTCH_SIGNALING gene set. To determine if epigenetic regulation of the Notch regulators contributes to their expression, we analyzed methylation levels of these factors using the TCGA HumanMethylation450 Array data. Notch receptors and ligands expression is generally associated with the tumor subtype, grade, and stage. Furthermore, we showed gene expression levels of most Notch factors were associated with DNA methylation rate. Modulating the expression levels of Notch receptors and effectors can be a potential therapeutic approach for breast cancer. As we outline herein, elucidating the novel prognostic and regulatory roles of Notch implicate this pathway as an essential mediator controlling breast cancer progression.
Collapse
Affiliation(s)
- Hassan Yousefi
- Biochemistry & Molecular Biology, Louisiana State University Health Science Center (LSUHSC), New Orleans, LA, USA
| | - Afshin Bahramy
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Narges Zafari
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahsa Rostamian Delavar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Khoa Nguyen
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA, USA
| | - Atousa Haghi
- Hematology Oncology and Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tahmineh Kandelouei
- Huntsman Cancer Institute, University of Utah School of Medicine, Salt Lake City, Utah, USA
| | - Cecilia Vittori
- Louisiana State University Health Sciences Center (LSUHSC), and Stanley S. Scott Cancer Center, New Orleans, LA, USA
| | - Parham Jazireian
- Department of Biology, University Campus 2, University of Guilan, Rasht, Iran
| | - Sajad Maleki
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Danyal Imani
- Department of Immunology, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amin Moshksar
- Interventional Radiology, University of Texas Medical Branch (UTMB), Galveston, TX, USA
| | - Amirreza Bitaraf
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box, Tehran, 14115-154, Iran
| | - Sadegh Babashah
- Department of Molecular Genetics, Faculty of Biological Sciences, Tarbiat Modares University, P.O. Box, Tehran, 14115-154, Iran.
| |
Collapse
|
5
|
Eljack S, David S, Chourpa I, Faggad A, Allard-Vannier E. Formulation of Lipid-Based Nanoparticles for Simultaneous Delivery of Lapatinib and Anti-Survivin siRNA for HER2+ Breast Cancer Treatment. Pharmaceuticals (Basel) 2022; 15:ph15121452. [PMID: 36558904 PMCID: PMC9784347 DOI: 10.3390/ph15121452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 11/13/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
In this work, lipid-based nanoparticles (LBNP) were designed to combine tyrosine kinase inhibitor (TKI) Lapatinib (LAPA) with siRNA directed against apoptosis inhibitor protein Survivin (siSurvivin) in an injectable form. This nanosystem is based on lipid nanocapsules (LNCs) coated with a cationic polymeric shell composed of chitosan grafted through a transacylation reaction. The hydrophobic LAPA is solubilized in the inner oily core, while hydrophilic siRNA is associated electrostatically onto the nanocarrier’s surface. The co-loaded LBNP showed a narrow size distribution (polydispersity index (PDI) < 0.3), a size of 130 nm, and a slightly positive zeta potential (+21 mV). LAPA and siRNA were loaded in LBNP at a high rate of >90% (10.6 mM) and 100% (4.6 µM), respectively. The siRNA-LAPA_LBNP was readily uptaken by the human epidermal growth factor receptor 2 overexpressed (HER2+) breast cancer cell line SK-BR-3. Moreover, the cytotoxicity studies confirmed that the blank chitosan decorated LBNP is not toxic to the cells with the tested concentrations, which correspond to LAPA concentrations from 1 to 10 µM, at different incubation times up to 96 h. Furthermore, siCtrl.-LAPA_LBNP had a more cytotoxic effect than Lapatinib salt, while siSurvivin-LAPA_LBNP had a significant synergistic cytotoxic effect compared to siCtrl.-LAPA_LBNP. All these findings suggested that the developed modified LBNP could potentiate anti-Survivin siRNA and LAPA anti-cancer activity.
Collapse
Affiliation(s)
- Sahar Eljack
- EA6295 Nanomédicaments et Nanosondes (NMNS), University of Tours, 37020 Tours, France
- Department of Pharmaceutics, Faculty of Pharmacy, University of Gezira, Wad Medani 21111, Sudan
| | - Stephanie David
- EA6295 Nanomédicaments et Nanosondes (NMNS), University of Tours, 37020 Tours, France
| | - Igor Chourpa
- EA6295 Nanomédicaments et Nanosondes (NMNS), University of Tours, 37020 Tours, France
| | - Areeg Faggad
- Department of Molecular Biology, National Cancer Institute, University of Gezira (NCI-UG), Wad Medani 21111, Sudan
| | - Emilie Allard-Vannier
- EA6295 Nanomédicaments et Nanosondes (NMNS), University of Tours, 37020 Tours, France
- Correspondence:
| |
Collapse
|
6
|
Tarantino P, Niman SM, Erick TK, Priedigkeit N, Harrison BT, Giordano A, Nakhlis F, Bellon JR, Parker T, Strauss S, Jin Q, King TA, Overmoyer BA, Curigliano G, Regan MM, Tolaney SM, Lynce F. HER2-low inflammatory breast cancer: Clinicopathologic features and prognostic implications. Eur J Cancer 2022; 174:277-286. [PMID: 36116830 DOI: 10.1016/j.ejca.2022.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/30/2022] [Accepted: 07/01/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND HER2)-low expression is a predictive biomarker for novel anti-HER2 antibody-drug conjugates. However, little is known about its clinical significance in inflammatory breast cancer (IBC). METHODS Patients diagnosed with HER2-negative IBC between December 1999 and December 2020 were identified from the Dana-Farber Cancer Institute IBC registry. Patients were divided into HER2-low (IHC 1+ or 2+/ISH-) and HER2-zero (IHC 0), comparing clinicopathologic features and disease outcomes between the two subgroups. RESULTS The study included 276 patients. Among patients with stage III (n = 209) and stage IV (n = 67) IBC, 54% and 39% had HER2-low tumours, respectively. Oestrogen receptor (ER)-expressing tumours were more common in patients with HER2-low versus HER2-zero stage III IBC (65% versus 38%, p < 0.01). Among stage III patients undergoing surgery (n = 182), pathologic complete response (pCR) rates were higher for HER2-zero versus HER2-low IBC (11% versus 6%, OR: 1.8, 95%CI:0.6-5.3), but minimal differences persisted when separately analysing pCR by ER status. Similar invasive disease-free survival (iDFS) outcomes were observed among ER-positive HER2-zero versus HER2-low IBC (48-month iDFS: 63% versus 63%, HR: 1.10, 95%CI:0.57-2.13) and ER-negative HER2-zero versus HER2-low IBC (48-month iDFS: 28% versus 25%, HR: 1.19, 95%CI:0.69-2.04). Differences in overall survival (OS) were small, both among ER-positive HER2-zero versus HER2-low IBC (48-month OS: 80% versus 81%, HR: 0.82, 95%CI:0.39-1.73) and ER-negative HER2-zero versus HER2-low IBC (48-month OS: 34% versus 47%, HR: 1.34, 95%CI: 0.74-2.41). CONCLUSIONS Marginal differences in clinicopathologic features and outcomes were observed in HER2-low versus HER2-zero IBC when controlling for ER status, not supporting the definition of HER2-low as a distinct subtype of IBC.
Collapse
Affiliation(s)
- Paolo Tarantino
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Centre, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Samuel M Niman
- Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Timothy K Erick
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Centre, Boston, MA, USA
| | - Nolan Priedigkeit
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Beth T Harrison
- Harvard Medical School, Boston, MA, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Antonio Giordano
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Centre, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Faina Nakhlis
- Breast Oncology Program, Dana-Farber Brigham Cancer Centre, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Jennifer R Bellon
- Breast Oncology Program, Dana-Farber Brigham Cancer Centre, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tonia Parker
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sarah Strauss
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Qingchun Jin
- Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Tari A King
- Breast Oncology Program, Dana-Farber Brigham Cancer Centre, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Breast Surgery, Department of Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Beth A Overmoyer
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Centre, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Giuseppe Curigliano
- Division of New Drugs and Early Drug Development, European Institute of Oncology, IRCCS, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Meredith M Regan
- Harvard Medical School, Boston, MA, USA; Data Sciences, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sara M Tolaney
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Centre, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Filipa Lynce
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Breast Oncology Program, Dana-Farber Brigham Cancer Centre, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
The Precise Detection of HER-2 Expression in Breast Cancer Cell via Au25 Probes. NANOMATERIALS 2022; 12:nano12060923. [PMID: 35335736 PMCID: PMC8950386 DOI: 10.3390/nano12060923] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 03/03/2022] [Accepted: 03/07/2022] [Indexed: 11/16/2022]
Abstract
Triple-negative breast cancer (TNBC) accounts for nearly one-quarter of all breast cancer cases, but effective targeted therapies for this disease remain elusive because TNBC cells lack the expression of the most common three receptors seen in other subtypes of breast cancers. The medium-term diagnosis of breast cancers is essential for development and prognosis. According to reports, patients with TNBC may be converted to a positive epidermal growth factor receptor 2(HER-2) after chemotherapy, and trastuzumab treatment will have a better prognosis. Therefore, it is important to accurately quantify the expression of HER-2 in breast cancer cells. Herein, we design a red fluorescent Au25 probe synthesized with BSA-biotin as the ligand, which is accurately quantified by HER-2 primary antibody-biotin using the avidin system. The quantitative detection of the expression of HER-2 in breast cancers is helpful for the companion diagnostic of breast cancer treatment and provides follow-up treatment.
Collapse
|
8
|
Bofinger R, Weitsman G, Evans R, Glaser M, Sander K, Allan H, Hochhauser D, Kalber TL, Årstad E, Hailes HC, Ng T, Tabor AB. Drug delivery, biodistribution and anti-EGFR activity: theragnostic nanoparticles for simultaneous in vivo delivery of tyrosine kinase inhibitors and kinase activity biosensors. NANOSCALE 2021; 13:18520-18535. [PMID: 34730152 PMCID: PMC8601123 DOI: 10.1039/d1nr02770k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 09/24/2021] [Indexed: 05/03/2023]
Abstract
In vivo delivery of small molecule therapeutics to cancer cells, assessment of the selectivity of administration, and measuring the efficacity of the drug in question at the molecule level, are important ongoing challenges in developing new classes of cancer chemotherapeutics. One approach that has the potential to provide targeted delivery, tracking of biodistribution and readout of efficacy, is to use multimodal theragnostic nanoparticles to deliver the small molecule therapeutic. In this paper, we report the development of targeted theragnostic lipid/peptide/DNA lipopolyplexes. These simultaneously deliver an inhibitor of the EGFR tyrosine kinase, and plasmid DNA coding for a Crk-based biosensor, Picchu-X, which when expressed in the target cells can be used to quantify the inhibition of EGFR in vivo in a mouse colorectal cancer xenograft model. Reversible bioconjugation of a known analogue of the tyrosine kinase inhibitor Mo-IPQA to a cationic peptide, and co-formulation with peptides containing both EGFR-binding and cationic sequences, allowed for good levels of inhibitor encapsulation with targeted delivery to LIM1215 colon cancer cells. Furthermore, high levels of expression of the Picchu-X biosensor in the LIM1215 cells in vivo allowed us to demonstrate, using fluorescence lifetime microscopy (FLIM)-based biosensing, that EGFR activity can be successfully suppressed by the tyrosine kinase inhibitor, released from the lipopolyplexes. Finally, we measured the biodistribution of lipopolyplexes containing 125I-labelled inhibitors and were able to demonstrate that the lipopolyplexes gave significantly higher drug delivery to the tumors compared with free drug.
Collapse
Affiliation(s)
- Robin Bofinger
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
| | - Gregory Weitsman
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK.
| | - Rachel Evans
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK.
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Matthias Glaser
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
- Centre for Radiopharmaceutical Chemistry, Kathleen Lonsdale Building, 5 Gower Place, London WC1E 6BS, UK
| | - Kerstin Sander
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
- Centre for Radiopharmaceutical Chemistry, Kathleen Lonsdale Building, 5 Gower Place, London WC1E 6BS, UK
| | - Helen Allan
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
| | - Daniel Hochhauser
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Erik Årstad
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
- Centre for Radiopharmaceutical Chemistry, Kathleen Lonsdale Building, 5 Gower Place, London WC1E 6BS, UK
| | - Helen C Hailes
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
| | - Tony Ng
- School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK.
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Alethea B Tabor
- Department of Chemistry, University College London, 20, Gordon Street, London WC1H 0AJ, UK.
| |
Collapse
|
9
|
Peptide Nanoparticle-Mediated Combinatorial Delivery of Cancer-Related siRNAs for Synergistic Anti-Proliferative Activity in Triple Negative Breast Cancer Cells. Pharmaceuticals (Basel) 2021; 14:ph14100957. [PMID: 34681181 PMCID: PMC8540820 DOI: 10.3390/ph14100957] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 09/15/2021] [Accepted: 09/21/2021] [Indexed: 12/16/2022] Open
Abstract
Triple negative breast cancer (TNBC) is one of the deadliest types of cancer for women of different age groups. Frequently this cancer does not respond to conservative treatment. Combinatorial RNAi can be suggested as an advanced approach to TNBC therapy. Due to the fact that TNBC cells overexpress chemokine receptor 4 we used modular L1 peptide-based nanoparticles modified with CXCR4 ligand for combinatorial delivery of siRNAs suppressing major transduction pathways. TNBC cell line MDA-MB-231 was used as a cellular model. Genes encoding the AQP3, CDC20, and COL4A2 proteins responsible for proliferative activity in TNBC cells were selected as RNAi targets. The siRNA binding ability of the carrier was studied at different charge ratios. The silencing specificity was demonstrated for all siRNAs studied. Alamar Blue proliferation assay has shown significant reduction in the anti-proliferative activity after combinatorial siRNA transfection compared to single siRNA delivery. The most significant synergistic effect has been demonstrated for combinatorial transfection of anti-COL4A2 and anti-CDC20 siRNAs what resulted in 1.5-2 fold inhibition of proliferation and migration of TNBC cells. Based on our findings, we have concluded that combinatorial treatment by CXCR4-ligand modified L1-polyplexes formed with AQP3, CDC20, and COL4A2 siRNAs effectively inhibits proliferation of TNBC cells and can be suggested as useful tool for RNAi-mediated cancer therapy.
Collapse
|
10
|
Crosby EJ, Acharya CR, Haddad AF, Rabiola CA, Lei G, Wei JP, Yang XY, Wang T, Liu CX, Wagner KU, Muller WJ, Chodosh LA, Broadwater G, Hyslop T, Shepherd JH, Hollern DP, He X, Perou CM, Chai S, Ashby BK, Vincent BG, Snyder JC, Force J, Morse MA, Lyerly HK, Hartman ZC. Stimulation of Oncogene-Specific Tumor-Infiltrating T Cells through Combined Vaccine and αPD-1 Enable Sustained Antitumor Responses against Established HER2 Breast Cancer. Clin Cancer Res 2020; 26:4670-4681. [PMID: 32732224 DOI: 10.1158/1078-0432.ccr-20-0389] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 04/17/2020] [Accepted: 06/25/2020] [Indexed: 12/15/2022]
Abstract
PURPOSE Despite promising advances in breast cancer immunotherapy, augmenting T-cell infiltration has remained a significant challenge. Although neither individual vaccines nor immune checkpoint blockade (ICB) have had broad success as monotherapies, we hypothesized that targeted vaccination against an oncogenic driver in combination with ICB could direct and enable antitumor immunity in advanced cancers. EXPERIMENTAL DESIGN Our models of HER2+ breast cancer exhibit molecular signatures that are reflective of advanced human HER2+ breast cancer, with a small numbers of neoepitopes and elevated immunosuppressive markers. Using these, we vaccinated against the oncogenic HER2Δ16 isoform, a nondriver tumor-associated gene (GFP), and specific neoepitopes. We further tested the effect of vaccination or anti-PD-1, alone and in combination. RESULTS We found that only vaccination targeting HER2Δ16, a driver of oncogenicity and HER2-therapeutic resistance, could elicit significant antitumor responses, while vaccines targeting a nondriver tumor-specific antigen or tumor neoepitopes did not. Vaccine-induced HER2-specific CD8+ T cells were essential for responses, which were more effective early in tumor development. Long-term tumor control of advanced cancers occurred only when HER2Δ16 vaccination was combined with αPD-1. Single-cell RNA sequencing of tumor-infiltrating T cells revealed that while vaccination expanded CD8 T cells, only the combination of vaccine with αPD-1 induced functional gene expression signatures in those CD8 T cells. Furthermore, we show that expanded clones are HER2-reactive, conclusively demonstrating the efficacy of this vaccination strategy in targeting HER2. CONCLUSIONS Combining oncogenic driver targeted vaccines with selective ICB offers a rational paradigm for precision immunotherapy, which we are clinically evaluating in a phase II trial (NCT03632941).
Collapse
Affiliation(s)
- Erika J Crosby
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Chaitanya R Acharya
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Anthony-Fayez Haddad
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Christopher A Rabiola
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Gangjun Lei
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Jun-Ping Wei
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Xiao-Yi Yang
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Tao Wang
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Cong-Xiao Liu
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina
| | - Kay U Wagner
- Department of Oncology, Wayne State University, Barbara Ann Karmanos Cancer Institute, Detroit, Michigan
| | - William J Muller
- Departments of Biochemistry and Medicine, Goodman Cancer Center, McGill University, Montreal, Quebec
| | - Lewis A Chodosh
- Department of Cancer Biology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gloria Broadwater
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Terry Hyslop
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina
| | - Jonathan H Shepherd
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Daniel P Hollern
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Xiaping He
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Charles M Perou
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Shengjie Chai
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, North Carolina
| | - Benjamin K Ashby
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, North Carolina
| | - Benjamin G Vincent
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina.,Department of Medicine, Division of Hematology/Oncology, University of North Carolina, Chapel Hill, North Carolina.,Curriculum in Bioinformatics and Computational Biology, University of North Carolina, Chapel Hill, North Carolina.,Computational Medicine Program, University of North Carolina, Chapel Hill, North Carolina
| | - Joshua C Snyder
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina.,Department of Cell Biology, Duke University, Durham, North Carolina
| | - Jeremy Force
- Department of Medicine, Duke University, Durham, North Carolina
| | - Michael A Morse
- Department of Medicine, Duke University, Durham, North Carolina
| | - Herbert K Lyerly
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina.,Department of Immunology, Duke University, Durham, North Carolina.,Department of Pathology, Duke University, Durham, North Carolina
| | - Zachary C Hartman
- Department of Surgery, Division of Surgical Sciences, Duke University, Durham North Carolina. .,Department of Pathology, Duke University, Durham, North Carolina
| |
Collapse
|
11
|
Du T, Shi Y, Xu S, Wan X, Sun H, Liu B. Long Non-Coding RNAs in Drug Resistance of Breast Cancer. Onco Targets Ther 2020; 13:7075-7087. [PMID: 32764993 PMCID: PMC7382578 DOI: 10.2147/ott.s255226] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 06/12/2020] [Indexed: 12/24/2022] Open
Abstract
Breast cancer (BC) is the most common cancer and the leading cause of death in women. Advances in early diagnosis and therapeutic strategies have decreased the mortality of BC and improved the prognosis of patients to some extent. However, the development of drug resistance has limited the success rate of systemic therapies. Long non-coding RNAs (lncRNAs) are involved in drug resistance in BC via various mechanisms, which contribute to a complex regulatory network. In this review, we summarize the latest findings on the mechanisms underlying drug resistance modulated by lncRNAs in BC. In addition, we discuss the potential clinical applications of lncRNAs as targeted molecular therapy against drug resistance in BC.
Collapse
Affiliation(s)
- Tonghua Du
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Ying Shi
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Shengnan Xu
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Xiaoyu Wan
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Haiyin Sun
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| | - Bin Liu
- Department of Breast Surgery, The Second Hospital of Jilin University, Changchun, Jilin, People's Republic of China
| |
Collapse
|
12
|
Chen AC, Xu R, Wang T, Wei J, Yang XY, Liu CX, Lei G, Lyerly HK, Heiland T, Hartman ZC. HER2-LAMP vaccines effectively traffic to endolysosomal compartments and generate enhanced polyfunctional T cell responses that induce complete tumor regression. J Immunother Cancer 2020; 8:jitc-2019-000258. [PMID: 32532838 PMCID: PMC7295440 DOI: 10.1136/jitc-2019-000258] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2020] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND The advent of immune checkpoint blockade antibodies has demonstrated that effective mobilization of T cell responses can cause tumor regression of metastatic cancers, although these responses are heterogeneous and restricted to certain histologic types of cancer. To enhance these responses, there has been renewed emphasis in developing effective cancer-specific vaccines to stimulate and direct T cell immunity to important oncologic targets, such as the oncogene human epidermal growth factor receptor 2 (HER2), expressed in ~20% of breast cancers (BCs). METHODS In our study, we explored the use of alternative antigen trafficking through use of a lysosome-associated membrane protein 1 (LAMP) domain to enhance vaccine efficacy against HER2 and other model antigens in both in vitro and in vivo studies. RESULTS We found that inclusion of this domain in plasmid vaccines effectively trafficked antigens to endolysosomal compartments, resulting in enhanced major histocompatibility complex (MHC) class I and II presentation. Additionally, this augmented the expansion/activation of antigen-specific CD4+ and CD8+ T cells and also led to elevated levels of antigen-specific polyfunctional CD8+ T cells. Significantly, vaccination with HER2-LAMP produced tumor regression in ~30% of vaccinated mice with established tumors in an endogenous model of metastatic HER2+ BC, compared with 0% of HER2-WT vaccinated mice. This therapeutic benefit is associated with enhanced tumor infiltration of activated CD4+ and CD8+ T cells. CONCLUSIONS These data demonstrate the potential of using LAMP-based endolysosomal trafficking as a means to augment the generation of polyfunctional, antigen-specific T cells in order to improve antitumor therapeutic responses using cancer antigen vaccines.
Collapse
Affiliation(s)
- Alan Chen Chen
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Renhuan Xu
- Department of R&D, Immunomic Therapeutics, Rockville, Maryland, USA
| | - Tao Wang
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Junping Wei
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Xiao-Yi Yang
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Cong-Xiao Liu
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Gangjun Lei
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA
| | - Herbert Kim Lyerly
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA.,Department of Pathology, Duke University, Durham, North Carolina, USA.,Department of Immunology, Duke University, Durham, NC, USA
| | - Teri Heiland
- Department of R&D, Immunomic Therapeutics, Rockville, Maryland, USA
| | - Zachary Conrad Hartman
- Department of Surgery, Duke University, Duke University, Durham, North Carolina, USA .,Department of Pathology, Duke University, Durham, North Carolina, USA
| |
Collapse
|
13
|
Ho-Pun-Cheung A, Bazin H, Boissière-Michot F, Mollevi C, Simony-Lafontaine J, Landas E, Bleuse JP, Chardès T, Prost JF, Pèlegrin A, Jacot W, Mathis G, Lopez-Crapez E. Quantification of HER1, HER2 and HER3 by time-resolved Förster resonance energy transfer in FFPE triple-negative breast cancer samples. Br J Cancer 2019; 122:397-404. [PMID: 31792349 PMCID: PMC7000684 DOI: 10.1038/s41416-019-0670-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2019] [Revised: 11/05/2019] [Accepted: 11/13/2019] [Indexed: 12/20/2022] Open
Abstract
Background Triple-negative breast cancer (TNBC) has a worse prognosis compared with other breast cancer subtypes, and biomarkers to identify patients at high risk of recurrence are needed. Here, we investigated the expression of human epidermal receptor (HER) family members in TNBC and evaluated their potential as biomarkers of recurrence. Methods We developed Time Resolved-Förster Resonance Energy Transfer (TR-FRET) assays to quantify HER1, HER2 and HER3 in formalin-fixed paraffin-embedded (FFPE) tumour tissues. After assessing the performance and precision of our assays, we quantified HER protein expression in 51 TNBC specimens, and investigated the association of their expression with relapse-free survival. Results The assays were quantitative, accurate, and robust. In TNBC specimens, HER1 levels ranged from ≈4000 to more than 2 million receptors per cell, whereas HER2 levels varied from ≈1000 to 60,000 receptors per cell. HER3 expression was very low (less than 5500 receptors per cell in all samples). Moderate HER2 expression was significantly associated with higher risk of recurrence (HR = 3.93; P = 0.003). Conclusions Our TR-FRET assays accurately quantify HER1, HER2 and HER3 in FFPE breast tumour specimens. Moderate HER2 expression may represent a novel prognostic marker in patients with TNBC.
Collapse
Affiliation(s)
- Alexandre Ho-Pun-Cheung
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | | | | | - Caroline Mollevi
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | | | | | | | - Thierry Chardès
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | | | - André Pèlegrin
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | - William Jacot
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France.,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France
| | | | - Evelyne Lopez-Crapez
- ICM, Institut régional du Cancer de Montpellier, Montpellier, France. .,Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM, Université de Montpellier, Institut régional du Cancer de Montpellier (ICM), Montpellier, France.
| |
Collapse
|
14
|
Adeleke S, Latifoltojar A, Sidhu H, Galazi M, Shah TT, Clemente J, Davda R, Payne HA, Chouhan MD, Lioumi M, Chua S, Freeman A, Rodriguez-Justo M, Coolen A, Vadgama S, Morris S, Cook GJ, Bomanji J, Arya M, Chowdhury S, Wan S, Haroon A, Ng T, Ahmed HU, Punwani S. Localising occult prostate cancer metastasis with advanced imaging techniques (LOCATE trial): a prospective cohort, observational diagnostic accuracy trial investigating whole-body magnetic resonance imaging in radio-recurrent prostate cancer. BMC Med Imaging 2019; 19:90. [PMID: 31730466 PMCID: PMC6858718 DOI: 10.1186/s12880-019-0380-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 09/13/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Accurate whole-body staging following biochemical relapse in prostate cancer is vital in determining the optimum disease management. Current imaging guidelines recommend various imaging platforms such as computed tomography (CT), Technetium 99 m (99mTc) bone scan and 18F-choline and recently 68Ga-PSMA positron emission tomography (PET) for the evaluation of the extent of disease. Such approach requires multiple hospital attendances and can be time and resource intensive. Recently, whole-body magnetic resonance imaging (WB-MRI) has been used in a single visit scanning session for several malignancies, including prostate cancer, with promising results, providing similar accuracy compared to the combined conventional imaging techniques. The LOCATE trial aims to investigate the application of WB-MRI for re-staging of patients with biochemical relapse (BCR) following external beam radiotherapy and brachytherapy in patients with prostate cancer. METHODS/DESIGN The LOCATE trial is a prospective cohort, multi-centre, non-randomised, diagnostic accuracy study comparing WB-MRI and conventional imaging. Eligible patients will undergo WB-MRI in addition to conventional imaging investigations at the time of BCR and will be asked to attend a second WB-MRI exam, 12-months following the initial scan. WB-MRI results will be compared to an enhanced reference standard comprising all the initial, follow-up imaging and non-imaging investigations. The diagnostic performance (sensitivity and specificity analysis) of WB-MRI for re-staging of BCR will be investigated against the enhanced reference standard on a per-patient basis. An economic analysis of WB-MRI compared to conventional imaging pathways will be performed to inform the cost-effectiveness of the WB-MRI imaging pathway. Additionally, an exploratory sub-study will be performed on blood samples and exosome-derived human epidermal growth factor receptor (HER) dimer measurements will be taken to investigate its significance in this cohort. DISCUSSION The LOCATE trial will compare WB-MRI versus the conventional imaging pathway including its cost-effectiveness, therefore informing the most accurate and efficient imaging pathway. TRIAL REGISTRATION LOCATE trial was registered on ClinicalTrial.gov on 18th of October 2016 with registration reference number NCT02935816.
Collapse
Affiliation(s)
- Sola Adeleke
- Centre for Medical Imaging, University College London, 2nd floor Charles Bell house, 43-45 Foley Street, London, W1W 7TS UK
| | - Arash Latifoltojar
- Centre for Medical Imaging, University College London, 2nd floor Charles Bell house, 43-45 Foley Street, London, W1W 7TS UK
| | - Harbir Sidhu
- Centre for Medical Imaging, University College London, 2nd floor Charles Bell house, 43-45 Foley Street, London, W1W 7TS UK
- Department of Radiology, University College London Hospital, London, 235 Euston Road, London, NW1 2BU UK
| | - Myria Galazi
- Molecular Oncology Group, University College London, Cancer Institute, Paul O’Gorman Building, 72 Huntley Street, London, WC1E 6DD UK
| | - Taimur T. Shah
- Division of Surgery and Interventional Science, University College London, 4th floor, 21 University Street, London, WC1E UK
- Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Department of Urology, Charing Cross Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Joey Clemente
- Centre for Medical Imaging, University College London, 2nd floor Charles Bell house, 43-45 Foley Street, London, W1W 7TS UK
| | - Reena Davda
- Oncology Department, University College London Hospital, 235 Euston Road, London, NW1 2BU UK
| | - Heather Ann Payne
- Oncology Department, University College London Hospital, 235 Euston Road, London, NW1 2BU UK
| | - Manil D. Chouhan
- Centre for Medical Imaging, University College London, 2nd floor Charles Bell house, 43-45 Foley Street, London, W1W 7TS UK
- Department of Radiology, University College London Hospital, London, 235 Euston Road, London, NW1 2BU UK
| | - Maria Lioumi
- Comprehensive Cancer Imaging Centre (CCIC), King’s College, London, New Hunt’s House, Guy’s Campus, London, SE1 1UL UK
| | - Sue Chua
- Department of Nuclear Medicine, The Royal Marsden Hospital NHS Foundation Trust, Down’s Road, Sutton, SM2 5PT UK
| | - Alex Freeman
- Histopathology Department, University College London Hospital, 4th Floor, Rockefeller Building University Street, London, WC1 6DE UK
| | - Manuel Rodriguez-Justo
- Histopathology Department, University College London Hospital, 4th Floor, Rockefeller Building University Street, London, WC1 6DE UK
| | - Anthony Coolen
- Institute for Mathematical and Molecular Biomedicine, King’s College London, Hodgkin Building, Guy’s Campus, London, SE1 1UL UK
| | - Sachin Vadgama
- Department of Applied Health Research, University College London, 1-19 Torrington Place, Fitzrovia, London, WC1E 7HB UK
| | - Steve Morris
- Department of Applied Health Research, University College London, 1-19 Torrington Place, Fitzrovia, London, WC1E 7HB UK
| | - Gary J. Cook
- Department of Cancer Imaging, School of Biomedical Engineering and Imaging Sciences, King’s College London, 4th Floor, Lambeth Wing St. Thomas’ Hospital, London, SE1 7EH UK
| | - Jamshed Bomanji
- Institute of Nuclear Medicine, University College London Hospital, 5th Floor Tower, 235 Euston Road, London, NW1 2BU UK
| | - Manit Arya
- Urology Department, University College Hospital, Westmoreland Street, 16-18 Westmoreland Street, London, W1G 8PH UK
| | - Simon Chowdhury
- Oncology Department, Guy’s and St. Thomas’ Hospital, Westminster Bridge road, Lambeth, London, SE1 7EH UK
| | - Simon Wan
- Institute of Nuclear Medicine, University College London Hospital, 5th Floor Tower, 235 Euston Road, London, NW1 2BU UK
| | - Athar Haroon
- Department of Nuclear Medicine, St Bartholomew’s Hospital, West Smithfield, London, EC1A 7BE UK
| | - Tony Ng
- Molecular Oncology Group, University College London, Cancer Institute, Paul O’Gorman Building, 72 Huntley Street, London, WC1E 6DD UK
| | - Hashim Uddin Ahmed
- Division of Surgery, Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, London, UK
- Urology Department, Imperial College Healthcare NHS Trust, London, W2 1NY UK
| | - Shonit Punwani
- Centre for Medical Imaging, University College London, 2nd floor Charles Bell house, 43-45 Foley Street, London, W1W 7TS UK
- Department of Radiology, University College London Hospital, London, 235 Euston Road, London, NW1 2BU UK
| |
Collapse
|
15
|
Huynh TK, Ho CY, Tsai CH, Wang CK, Chen YJ, Bau DT, Tu CY, Li TS, Huang WC. Proteasome Inhibitors Suppress ErbB Family Expression through HSP90-Mediated Lysosomal Degradation. Int J Mol Sci 2019; 20:ijms20194812. [PMID: 31569723 PMCID: PMC6801459 DOI: 10.3390/ijms20194812] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2019] [Revised: 09/19/2019] [Accepted: 09/21/2019] [Indexed: 12/31/2022] Open
Abstract
Although dual EGFR/HER2 tyrosine kinase inhibitor lapatinib has provided effective clinical benefits for HER2-positive breast cancer patients, acquired resistance to this drug remains a major concern. Thus, the development of alternative therapeutic strategies is urgently needed for patients who failed lapatinib treatment. Proteasome inhibitors have been reported to possess high anti-tumor activity to breast cancer cells. Therefore, this study aims to examine whether and how proteasome inhibitor bortezomib can overcome lapatinib resistance. Treatments with several proteasome inhibitors, including Bortezomib, MG132, and proteasome inhibitor I (PSI), as well as the viabilities of both HER2-positive breast cancer cell lines and their lapatinib-resistant clones, were inhibited. Importantly, the expressions of ErbB family were downregulated at both transcriptional and translational levels. Also, our results further indicated that proteasome inhibitors decreased ErbB family expression through lysosomal degradation pathway in a heat shock protein 90 (HSP90)-dependent manner. In this study, our data supported a potential approach to overcome the acquired resistance of HER2-overexpressing breast cancer patients to lapatinib using proteasome inhibitors.
Collapse
Affiliation(s)
- Thanh Kieu Huynh
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404, Taiwan.
| | - Chien-Yi Ho
- Department of Biomedical Imaging and Radiological Science, China Medical University, Taichung 404, Taiwan.
- Department of Family Medicine, China Medical University Hsinchu Hospital, Hsinchu 302, Taiwan.
- Physical Examination Center, China Medical University Hsinchu Hospital, Hsinchu 302, Taiwan.
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu 302, Taiwan.
| | - Chi-Hua Tsai
- Graduate Institute of Cancer Biology, China Medical University, Taichung 404, Taiwan.
| | - Chien-Kuo Wang
- Department of Biotechnology, Asia University, Taichung 413, Taiwan.
| | - Yun-Ju Chen
- Department of Medical Research, E-Da Hospital, Kaohsiung 824, Taiwan.
- School of Medicine for International Students, I-Shou University, Kaohsiung 824, Taiwan.
- Department of Pharmacy, E-Da Hospital, Kaohsiung 824, Taiwan.
| | - Da-Tian Bau
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404, Taiwan.
- Terry Fox Cancer Research Laboratory, Translational Medicine Research Center, China Medical University Hospital, Taichung 404, Taiwan.
| | - Chih-Yen Tu
- School of Medicine, College of Medicine, China Medical University, Taichung 404, Taiwan.
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung 404, Taiwan.
| | - Tzong-Shiun Li
- Department of Plastic Surgery, Show Chwan Memorial Hospital, Changhua 500, Taiwan.
- Innovation Research Center, Show Chwan Health Care System, Changhua 500, Taiwan.
| | - Wei-Chien Huang
- Graduate Institute of Biomedical Science, China Medical University, Taichung 404, Taiwan.
- Graduate Institute of Cancer Biology, China Medical University, Taichung 404, Taiwan.
- Department of Biotechnology, Asia University, Taichung 413, Taiwan.
- The Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University, Taichung 404, Taiwan.
- Center for Molecular Medicine, China Medical University Hospital, Taichung 404, Taiwan.
- Drug Development Center, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
16
|
Protease Nexin I is a feedback regulator of EGF/PKC/MAPK/EGR1 signaling in breast cancer cells metastasis and stemness. Cell Death Dis 2019; 10:649. [PMID: 31501409 PMCID: PMC6733841 DOI: 10.1038/s41419-019-1882-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Revised: 08/07/2019] [Accepted: 08/26/2019] [Indexed: 01/18/2023]
Abstract
Breast cancer is the most prevalent cancer in women worldwide, which remains incurable once metastatic. Breast cancer stem cells (BCSCs) are a small subset of breast cancer cells, which are the radical cause of drug resistance, tumor relapse, and metastasis in breast cancer. The extracellular serine protease inhibitor serpinE2, also named protease nexin-1 (PN-1), contributes to enhanced metastasis of cancer cells mainly by remodeling the tumor matrix. In this study, we found that PN-1 was up-regulated in breast cancer, which promoted cell invasion, migration and stemness. Furthermore, by using specific inhibitors, we discovered that epidermal growth factor (EGF) up-regulated PN-1 in breast cancer cells through cascade activation of epidermal growth factor receptor (EGFR) to the activation of protein kinase Cδ (PKCδ), mitogen-activated protein kinase (MEK) and extracellular signal-related kinase (ERK), which finally led to the up-regulation of early growth response protein 1 (EGR1). Moreover, EGF signaling was further activated as a feedback of PN-1 up-regulation through PN-1 blocking HtrA1. Taken together, our findings revealed a novel signaling axis that up-regulated PN-1 expression in breast cancer cells, and the new mechanism of PN-1-promoted breast cancer metastasis, which may provide new insights into identifying novel therapeutic targets for breast cancer.
Collapse
|
17
|
Jiang C, Wu S, Jiang L, Gao Z, Li X, Duan Y, Li N, Sun T. Network-based approach to identify biomarkers predicting response and prognosis for HER2-negative breast cancer treatment with taxane-anthracycline neoadjuvant chemotherapy. PeerJ 2019; 7:e7515. [PMID: 31534839 PMCID: PMC6730536 DOI: 10.7717/peerj.7515] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 07/18/2019] [Indexed: 11/20/2022] Open
Abstract
OBJECTIVE This study aims to identify effective gene networks and biomarkers to predict response and prognosis for HER2-negative breast cancer patients who received sequential taxane-anthracycline neoadjuvant chemotherapy. MATERIALS AND METHODS Transcriptome data of training dataset including 310 HER2-negative breast cancer who received taxane-anthracycline treatment and an independent validation set with 198 samples were analyzed by weighted gene co-expression network analysis (WGCNA) approach in R language. Gene ontology (GO) terms and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis were performed for the selected genes. Module-clinical trait relationships were analyzed to explore the genes and pathways that associated with clinicopathological parameters. Log-rank tests and COX regression were used to identify the prognosis-related genes. RESULTS We found a significant correlation of an expression module with distant relapse-free survival (HR = 0.213, 95% CI [0.131-0.347], P = 4.80E-9). This blue module contained genes enriched in biological process of hormone levels regulation, reproductive system, response to estradiol, cell growth and mammary gland development as well as pathways including estrogen, apelin, cAMP, the PPAR signaling pathway and fatty acid metabolism. From this module, we further screened and validated six hub genes (CA12, FOXA1, MLPH, XBP1, GATA3 and MAGED2), the expression of which were significantly associated with both better chemotherapeutic response and favorable survival for BC patients. CONCLUSION We used WGCNA approach to reveal a gene network that regulate HER2-negative breast cancer treatment with taxane-anthracycline neoadjuvant chemotherapy, which enriched in pathways of estrogen signaling, apelin signaling, cAMP signaling, the PPAR signaling pathway and fatty acid metabolism. In addition, genes of CA12, FOXA1, MLPH, XBP1, GATA3 and MAGED2 might serve as novel biomarkers predicting chemotherapeutic response and prognosis for HER2-negative breast cancer.
Collapse
Affiliation(s)
- Cui Jiang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Shuo Wu
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Lei Jiang
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Zhichao Gao
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Xiaorui Li
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Yangyang Duan
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Na Li
- Institute of Translational Medicine, China Medical University, Shenyang, Liaoning, China
| | - Tao Sun
- Department of Medical Oncology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| |
Collapse
|
18
|
Zhang W, Yu G, Zhang Y, Tang F, Lv J, Tian G, Zhang Y, Liu J, Mi J, Zhang J. Quantitative Dot Blot (QDB) as a universal platform for absolute quantification of tissue biomarkers. Anal Biochem 2019; 576:42-47. [PMID: 30974091 DOI: 10.1016/j.ab.2019.04.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/27/2019] [Accepted: 04/03/2019] [Indexed: 12/22/2022]
Abstract
To circumvent the limitations associated with sandwich ELISA for tissue biomarker quantitation, Quantitative Dot Blot method (QDB) was proposed using antibodies clinically validated for immunohistochemistry (IHC), as this method requires only one primary antibody in the analysis. The protein levels of four breast cancer tissue biomarkers, including Estrogen Receptor (ER), Progesterone Receptor (PR), Ki67 and Her2, were absolutely quantitated successfully in 190 frozen breast tissue biopsies, and the results were further verified with provided IHC results. We propose QDB method as an alternative platform to Sandwich ELISA for absolute quantitation of tissue biomarkers with significantly reduced developing effort and time.
Collapse
Affiliation(s)
- Wenfeng Zhang
- Yantai Quanticision Diagnostics Inc., A Division of Quanticision Diagnostics, Inc. (US), Yantai Hi-tech District, Shandong, 264670, PR China
| | - Guohua Yu
- Laboratory of Molecular Pathology, Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, PR China
| | - Yunyun Zhang
- Yantai Quanticision Diagnostics Inc., A Division of Quanticision Diagnostics, Inc. (US), Yantai Hi-tech District, Shandong, 264670, PR China
| | - Fangrong Tang
- Yantai Quanticision Diagnostics Inc., A Division of Quanticision Diagnostics, Inc. (US), Yantai Hi-tech District, Shandong, 264670, PR China
| | - Jiahong Lv
- Yantai Quanticision Diagnostics Inc., A Division of Quanticision Diagnostics, Inc. (US), Yantai Hi-tech District, Shandong, 264670, PR China
| | - Geng Tian
- Medicine and Pharmarcy Research Center, Precision Medicine Division, Binzhou Medical University, Laishan District, Shandong, 264003, PR China
| | - Yuan Zhang
- Medicine and Pharmarcy Research Center, Precision Medicine Division, Binzhou Medical University, Laishan District, Shandong, 264003, PR China
| | - Jing Liu
- Laboratory of Molecular Pathology, Department of Pathology, Affiliated Yantai Yuhuangding Hospital, Qingdao University, PR China
| | - Jia Mi
- Medicine and Pharmarcy Research Center, Precision Medicine Division, Binzhou Medical University, Laishan District, Shandong, 264003, PR China.
| | - Jiandi Zhang
- Yantai Quanticision Diagnostics Inc., A Division of Quanticision Diagnostics, Inc. (US), Yantai Hi-tech District, Shandong, 264670, PR China; Medicine and Pharmarcy Research Center, Precision Medicine Division, Binzhou Medical University, Laishan District, Shandong, 264003, PR China.
| |
Collapse
|
19
|
Comparison of Antibodies for Immunohistochemistry-based Detection of HER3 in Breast Cancer. Appl Immunohistochem Mol Morphol 2019; 26:212-219. [PMID: 27389555 DOI: 10.1097/pai.0000000000000406] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND Growth factor receptor HER3 (ErbB3) lacks standardized immunohistochemistry (IHC)-based methods for formalin-fixed paraffin-embedded (FFPE) tissue samples. We compared 4 different anti-HER3 antibodies to explain the differences found in the staining results reported in the literature. MATERIALS AND METHODS Four commercial HER3 antibodies were tested on FFPE samples including mouse monoclonal antibody clones, DAK-H3-IC and RTJ1, rabbit monoclonal antibody clone SP71, and rabbit polyclonal antibody (SAB4500793). Membranous and cytoplasmic staining patterns were analyzed and scored as 0, 1+, or 2+ according to the intensity of the staining and completeness of membranous and cytoplasmic staining. A large collection of HER2-amplified breast cancers (n=177) was stained with the best performing HER3 antibody. The breast cancer cell line, MDA-453, and human prostate tissue were used as positive controls. IHC results were confirmed by analysis of flow cytometry performed on breast cancer cell lines. Staining results of FFPE samples were compared with samples fixed with an epitope-sensitive fixative (PAXgene). RESULTS Clear circumferential cell membrane staining was found only with the HER3 antibody clone DAK-H3-IC. Other antibodies (RTJ1, SP71, and polyclonal) yielded uncertain and nonreproducible staining results. In addition to cell membrane staining, DAK-H3-IC was also localized to the cytoplasm, but no nuclear staining was observed. In HER2-amplified breast cancers, 80% of samples were classified as 1+ or 2+ according to the HER3 staining on the cell membrane. The results from FFPE cell line samples were comparable to those obtained from unfixed cells in flow cytometry. IHC conducted on FFPE samples and on PAXgene-fixed samples showed equivalent results. CONCLUSIONS We conclude that IHC with the monoclonal antibody, DAK-H3-IC, on FFPE samples is a reliable staining method for use in translational research. Assessment of membranous HER3 expression may be clinically relevant in selecting patients who may most benefit from pertuzumab or other novel anti-HER3 therapies.
Collapse
|
20
|
Wege AK, Chittka D, Buchholz S, Klinkhammer-Schalke M, Diermeier-Daucher S, Zeman F, Ortmann O, Brockhoff G. HER4 expression in estrogen receptor-positive breast cancer is associated with decreased sensitivity to tamoxifen treatment and reduced overall survival of postmenopausal women. Breast Cancer Res 2018; 20:139. [PMID: 30458882 PMCID: PMC6247692 DOI: 10.1186/s13058-018-1072-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 11/01/2018] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND The sensitivity of estrogen receptor-positive breast cancers to tamoxifen treatment varies considerably, and the molecular mechanisms affecting the response rates are manifold. The human epidermal growth factor receptor-related receptor HER2 is known to trigger intracellular signaling cascades that modulate the activity of coregulators of the estrogen receptor which, in turn, reduces the cell sensitivity to tamoxifen treatment. However, the impact of HER2-related receptor tyrosine kinases HER1, HER3, and, in particular, HER4 on endocrine treatment is largely unknown. METHODS Here, we retrospectively evaluated the importance of HER4 expression on the outcome of tamoxifen- and aromatase inhibitor-treated estrogen receptor-positive breast cancer patients (n = 258). In addition, we experimentally analyzed the efficiency of tamoxifen treatment as a function of HER4 co-expression in vitro. RESULTS We found a significantly improved survival in tamoxifen-treated postmenopausal breast cancer patients in the absence of HER4 compared with those with pronounced HER4 expression. In accordance with this finding, the sensitivity to tamoxifen treatment of estrogen and HER4 receptor-positive ZR-75-1 breast cancer cells can be significantly enhanced by HER4 knockdown. CONCLUSION We suggest an HER4/estrogen receptor interaction that impedes tamoxifen binding to the estrogen receptor and reduces treatment efficiency. Whether the sensitivity to tamoxifen treatment can be enhanced by anti-HER4 targeting needs to be prospectively evaluated.
Collapse
Affiliation(s)
- Anja Kathrin Wege
- Clinic of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
| | - Dominik Chittka
- Clinic of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany.,Department of Nephrology, University Hospital Regensburg, Regensburg, Germany
| | - Stefan Buchholz
- Clinic of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
| | | | | | - Florian Zeman
- Center for Clinical Studies, University Hospital Regensburg, Regensburg, Germany
| | - Olaf Ortmann
- Clinic of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany
| | - Gero Brockhoff
- Clinic of Gynecology and Obstetrics, University Medical Center Regensburg, Regensburg, Germany.
| |
Collapse
|
21
|
Pecero ML, Salvador-Bofill J, Molina-Pinelo S. Long non-coding RNAs as monitoring tools and therapeutic targets in breast cancer. Cell Oncol (Dordr) 2018; 42:1-12. [DOI: 10.1007/s13402-018-0412-6] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/02/2018] [Indexed: 12/31/2022] Open
|
22
|
Monypenny J, Milewicz H, Flores-Borja F, Weitsman G, Cheung A, Chowdhury R, Burgoyne T, Arulappu A, Lawler K, Barber PR, Vicencio JM, Keppler M, Wulaningsih W, Davidson SM, Fraternali F, Woodman N, Turmaine M, Gillett C, Franz D, Quezada SA, Futter CE, Von Kriegsheim A, Kolch W, Vojnovic B, Carlton JG, Ng T. ALIX Regulates Tumor-Mediated Immunosuppression by Controlling EGFR Activity and PD-L1 Presentation. Cell Rep 2018; 24:630-641. [PMID: 30021161 PMCID: PMC6077252 DOI: 10.1016/j.celrep.2018.06.066] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 04/23/2018] [Accepted: 06/15/2018] [Indexed: 12/25/2022] Open
Abstract
The immunosuppressive transmembrane protein PD-L1 was shown to traffic via the multivesicular body (MVB) and to be released on exosomes. A high-content siRNA screen identified the endosomal sorting complexes required for transport (ESCRT)-associated protein ALIX as a regulator of both EGFR activity and PD-L1 surface presentation in basal-like breast cancer (BLBC) cells. ALIX depletion results in prolonged and enhanced stimulation-induced EGFR activity as well as defective PD-L1 trafficking through the MVB, reduced exosomal secretion, and its redistribution to the cell surface. Increased surface PD-L1 expression confers an EGFR-dependent immunosuppressive phenotype on ALIX-depleted cells. An inverse association between ALIX and PD-L1 expression was observed in human breast cancer tissues, while an immunocompetent mouse model of breast cancer revealed that ALIX-deficient tumors are larger and show an increased immunosuppressive environment. Our data suggest that ALIX modulates immunosuppression through regulation of PD-L1 and EGFR and may, therefore, present a diagnostic and therapeutic target for BLBC.
Collapse
Affiliation(s)
- James Monypenny
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK
| | - Hanna Milewicz
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK
| | - Fabian Flores-Borja
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK; KCL Breast Cancer Now Research Unit, Department of Research Oncology, Guy's Hospital, King's College London, London SE1 9RT, UK
| | - Gregory Weitsman
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK
| | - Anthony Cheung
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK; KCL Breast Cancer Now Research Unit, Department of Research Oncology, Guy's Hospital, King's College London, London SE1 9RT, UK
| | - Ruhe Chowdhury
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK; Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, Great Maze Pond, London, UK
| | - Thomas Burgoyne
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Appitha Arulappu
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK
| | - Katherine Lawler
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK; Institute for Mathematical and Molecular Biomedicine, King's College London, Guy's Medical School Campus, London SE1 1UL, UK
| | - Paul R Barber
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK; UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Jose M Vicencio
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Melanie Keppler
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK
| | - Wahyu Wulaningsih
- Cancer Epidemiology Group, Division of Cancer Studies, King's College London, London, UK
| | - Sean M Davidson
- Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London WC1E 6HX, UK
| | - Franca Fraternali
- Bioinformatics and Computational Biology, Randall Division, King's College London, Guy's Medical School Campus, London SE1 1UL, UK
| | - Natalie Woodman
- KHP Cancer Biobank, King's College London, Innovation Hub, Guy's Cancer Centre, London SE1 9RT, UK
| | - Mark Turmaine
- Division of Biosciences, University College London, Gower Street, London WC1E 6BT, UK
| | - Cheryl Gillett
- KHP Cancer Biobank, King's College London, Innovation Hub, Guy's Cancer Centre, London SE1 9RT, UK
| | - Dafne Franz
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Sergio A Quezada
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK
| | - Clare E Futter
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Alex Von Kriegsheim
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland
| | - Walter Kolch
- Systems Biology Ireland, University College Dublin, Belfield, Dublin 4, Ireland; Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland; School of Medicine, University College Dublin, Belfield, Dublin 4, Ireland
| | - Borivoj Vojnovic
- Department of Oncology, Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, University of Oxford, Oxford OX3 7DQ, UK
| | - Jeremy G Carlton
- Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Hospital, Great Maze Pond, London, UK; Organelle Dynamics Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK.
| | - Tony Ng
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer and Pharmaceutical Sciences, King's College London, Guy's Medical School Campus, London SE1 1UL, UK; KCL Breast Cancer Now Research Unit, Department of Research Oncology, Guy's Hospital, King's College London, London SE1 9RT, UK; UCL Cancer Institute, Paul O'Gorman Building, University College London, London WC1E 6DD, UK.
| |
Collapse
|
23
|
Abstract
There are two aspects of immunohistochemistry (IHC) that are relevant to practicing pathologist: (1) understanding of IHC biomarker panels that are suitable for diagnostic, prognostic and predictive testing, and (2) understanding of IHC quality assurance (QA), which makes sure that the tests in these panels work as they should. The two aspects are closely linked together and call for collaborative approach between pathologists and IHC laboratory technologists as both need to be involved in developing and maintaining IHC biomarkers that are "fit-for-purpose". This article reviews the most current IHC QA concepts that are imminently material to practicing pathologists with emphasis on challenges that are specific to endocrine pathology.
Collapse
Affiliation(s)
- Emina Emilia Torlakovic
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Saskatchewan, and Saskatchewan Health Authority, Saskatoon, Canada.
- Department of Pathology and Laboratory Medicine, Royal University Hospital, 103 Hospital Drive, Saskatoon, SK, S7N 0W8, Canada.
| |
Collapse
|
24
|
Baldassarre T, Truesdell P, Craig AW. Endophilin A2 promotes HER2 internalization and sensitivity to trastuzumab-based therapy in HER2-positive breast cancers. Breast Cancer Res 2017; 19:110. [PMID: 28974266 PMCID: PMC5627411 DOI: 10.1186/s13058-017-0900-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 08/30/2017] [Indexed: 12/13/2022] Open
Abstract
Background Human epidermal growth factor receptor-2 (HER2) is amplified and a clinical target in a subset of human breast cancers with high rates of metastasis. Targeted therapies involving the antibody trastuzumab and trastuzumab-emtansine (T-DM1) have greatly improved outcomes for HER2-positive (HER2+) breast cancer patients. However, resistance to these targeted therapies can develop and limit their efficacy. Here, we test the involvement of the endocytic adaptor protein endophilin A2 (Endo II) in HER2+ breast cancer models, and their responses to treatments with trastuzumab and T-DM1. Methods Endo II expression in human breast tumors and lymph node metastases were analyzed by immunohistochemistry. Stable silencing of Endo II was achieved in HER2+ cancer cell lines (SK-BR-3 and HCC1954) to test Endo II effects on HER2 levels, localization and signaling, cell motility and tumor metastasis. The effects of Endo II silencing on the responses of HER2+ cancer cells to trastuzumab or T-DM1 treatments were tested using real-time cell motility and cytotoxicity assays. Results High Endo II protein expression was detected in HER2-positive tumors, and was linked to worse overall survival in node-positive HER2+ breast cancers at the mRNA level. Stable silencing of Endo II in HER2+ cell lines led to elevated levels of HER2 on the cell surface, impaired epidermal growth factor-induced HER2 internalization, and reduced signaling to downstream effector kinases Akt and Erk. Endo II silencing also led to decreased migration and invasion of HER2+ cancer cells in vitro, and impaired lung seeding following tail vein injection in mice. In addition, Endo II silencing also impaired HER2 internalization in response to Trastuzumab, and led to reduced cytotoxicity response in HER2+ cancer cells treated with T-DM1. Conclusions Our study provides novel evidence of Endo II function in HER2+ cancer cell motility and trafficking of HER2 that relates to effective treatments with trastuzumab or T-DM1. Thus, differential expression of Endo II may relate to sensitivity or resistance to trastuzumab-based therapies for HER2+ cancers. Electronic supplementary material The online version of this article (doi:10.1186/s13058-017-0900-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tomas Baldassarre
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Peter Truesdell
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada.,Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Ontario, Canada
| | - Andrew W Craig
- Department of Biomedical and Molecular Sciences, Queen's University, Kingston, Ontario, Canada. .,Cancer Biology & Genetics Division, Queen's Cancer Research Institute, Kingston, Ontario, Canada.
| |
Collapse
|
25
|
Broughton MN, Westgaard A, Paus E, Øijordsbakken M, Henanger KJ, Naume B, Bjøro T. Specific antibodies and sensitive immunoassays for the human epidermal growth factor receptors (HER2, HER3, and HER4). Tumour Biol 2017; 39:1010428317707436. [PMID: 28653892 DOI: 10.1177/1010428317707436] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
The use of trastuzumab in patients with breast cancer that overexpresses human epidermal growth factor receptor 2 has significantly improved treatment outcomes. However, a substantial proportion of this patient group still experiences progression of the disease after receiving the drug. Evaluation of the changes in expression of the human epidermal growth factor receptors could be of interest. Monoclonal antibodies against the extracellular domain of the human growth factor receptors, 2, 3, and 4, have been raised, and specific and sensitive immunoassays have been established. Sera from healthy individuals (Nordic Reference Interval Project and Database) were analyzed in the human epidermal growth factor receptor 2 assay (N = 805) and the human epidermal growth factor receptor 3 and 4 assays (N = 114), and reference limits were calculated. In addition, sera from 208 individual patients with breast cancer were tested in all three assays. Finally, the human epidermal growth factor receptor 2 assay was compared with a chemiluminescent immunoassay for serum human epidermal growth factor receptor 2/neu. Reference values were as follows: human epidermal growth factor receptor 2, <2.5 µg/L; human epidermal growth factor receptor 3, <2.8 µg/L; and human epidermal growth factor receptor 4, <1.8 µg/L. There were significant differences in human epidermal growth factor receptor 2 and human epidermal growth factor receptor 3 serum levels between the patients with tissue human epidermal growth factor receptor 2-positive and tissue human epidermal growth factor receptor 2-negative ( p = 0.0026, p = 0.000011) tumors, but not in the serum levels of human epidermal growth factor receptor 4 ( p = 0.054). There was good agreement between the in-house human epidermal growth factor receptor 2 assay and the chemiluminescent immunoassay. Our new specific antibodies for all the three human epidermal growth factor receptors may prove valuable in the development of novel anti-human epidermal growth factor receptor targeted therapies with sensitive immunoassays for measuring serum levels of the respective targets and in monitoring established treatment.
Collapse
Affiliation(s)
| | - Arne Westgaard
- 2 Department of Oncology, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
| | - Elisabeth Paus
- 1 Department of Medical Biochemistry, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
| | - Miriam Øijordsbakken
- 1 Department of Medical Biochemistry, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
| | - Karoline J Henanger
- 1 Department of Medical Biochemistry, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
| | - Bjørn Naume
- 2 Department of Oncology, Radiumhospitalet, Oslo University Hospital, Oslo, Norway.,3 Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Trine Bjøro
- 1 Department of Medical Biochemistry, Radiumhospitalet, Oslo University Hospital, Oslo, Norway.,3 Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
26
|
Weitsman G, Mitchell NJ, Evans R, Cheung A, Kalber TL, Bofinger R, Fruhwirth GO, Keppler M, Wright ZVF, Barber PR, Gordon P, de Koning T, Wulaningsih W, Sander K, Vojnovic B, Ameer-Beg S, Lythgoe M, Arnold JN, Årstad E, Festy F, Hailes HC, Tabor AB, Ng T. Detecting intratumoral heterogeneity of EGFR activity by liposome-based in vivo transfection of a fluorescent biosensor. Oncogene 2017; 36:3618-3628. [PMID: 28166195 PMCID: PMC5421598 DOI: 10.1038/onc.2016.522] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/12/2016] [Accepted: 12/21/2016] [Indexed: 12/20/2022]
Abstract
Despite decades of research in the epidermal growth factor receptor (EGFR) signalling field, and many targeted anti-cancer drugs that have been tested clinically, the success rate for these agents in the clinic is low, particularly in terms of the improvement of overall survival. Intratumoral heterogeneity is proposed as a major mechanism underlying treatment failure of these molecule-targeted agents. Here we highlight the application of fluorescence lifetime microscopy (FLIM)-based biosensing to demonstrate intratumoral heterogeneity of EGFR activity. For sensing EGFR activity in cells, we used a genetically encoded CrkII-based biosensor which undergoes conformational changes upon tyrosine-221 phosphorylation by EGFR. We transfected this biosensor into EGFR-positive tumour cells using targeted lipopolyplexes bearing EGFR-binding peptides at their surfaces. In a murine model of basal-like breast cancer, we demonstrated a significant degree of intratumoral heterogeneity in EGFR activity, as well as the pharmacodynamic effect of a radionuclide-labeled EGFR inhibitor in situ. Furthermore, a significant correlation between high EGFR activity in tumour cells and macrophage-tumour cell proximity was found to in part account for the intratumoral heterogeneity in EGFR activity observed. The same effect of macrophage infiltrate on EGFR activation was also seen in a colorectal cancer xenograft. In contrast, a non-small cell lung cancer xenograft expressing a constitutively active EGFR conformational mutant exhibited macrophage proximity-independent EGFR activity. Our study validates the use of this methodology to monitor therapeutic response in terms of EGFR activity. In addition, we found iNOS gene induction in macrophages that are cultured in tumour cell-conditioned media as well as an iNOS activity-dependent increase in EGFR activity in tumour cells. These findings point towards an immune microenvironment-mediated regulation that gives rise to the observed intratumoral heterogeneity of EGFR signalling activity in tumour cells in vivo.
Collapse
Affiliation(s)
- G Weitsman
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - N J Mitchell
- Department of Chemistry, University College London, London, UK
| | - R Evans
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - A Cheung
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
- Breast Cancer Now Research Unit, King’s College London, London, UK
| | - T L Kalber
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, UK
| | - R Bofinger
- Department of Chemistry, University College London, London, UK
| | - G O Fruhwirth
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - M Keppler
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - Z V F Wright
- Department of Chemistry, University College London, London, UK
| | - P R Barber
- Gray Laboratories, Department of Oncology, Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - P Gordon
- Breast Cancer Now Research Unit, King’s College London, London, UK
| | - T de Koning
- Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - W Wulaningsih
- Cancer Epidemiology Group, Division of Cancer Studies, King’s College London, London, UK
| | - K Sander
- Institute of Nuclear Medicine, University College London, London, UK
| | - B Vojnovic
- Gray Laboratories, Department of Oncology, Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, Oxford, UK
| | - S Ameer-Beg
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - M Lythgoe
- UCL Centre for Advanced Biomedical Imaging, Division of Medicine, University College London, London, UK
| | - J N Arnold
- Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
| | - E Årstad
- Institute of Nuclear Medicine, University College London, London, UK
| | - F Festy
- King’s College London Dental Institute, Tissue Engineering and Biophotonics, Guy’s Hospital Campus, London, UK
| | - H C Hailes
- Department of Chemistry, University College London, London, UK
| | - A B Tabor
- Department of Chemistry, University College London, London, UK
| | - T Ng
- Richard Dimbleby Department of Cancer Research, Randall Division & Division of Cancer Studies, Kings College London, Guy’s Medical School Campus, London, UK
- Breast Cancer Now Research Unit, King’s College London, London, UK
- UCL Cancer Institute, Paul O’Gorman Building, University College London, London, UK
| |
Collapse
|
27
|
Hierro C, Alsina M, Sánchez M, Serra V, Rodon J, Tabernero J. Targeting the fibroblast growth factor receptor 2 in gastric cancer: promise or pitfall? Ann Oncol 2017; 28:1207-1216. [DOI: 10.1093/annonc/mdx081] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
28
|
Masoud V, Pagès G. Targeted therapies in breast cancer: New challenges to fight against resistance. World J Clin Oncol 2017; 8:120-134. [PMID: 28439493 PMCID: PMC5385433 DOI: 10.5306/wjco.v8.i2.120] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 09/16/2016] [Accepted: 10/17/2016] [Indexed: 02/06/2023] Open
Abstract
Breast cancer is the most common type of cancer found in women and today represents a significant challenge to public health. With the latest breakthroughs in molecular biology and immunotherapy, very specific targeted therapies have been tailored to the specific pathophysiology of different types of breast cancers. These recent developments have contributed to a more efficient and specific treatment protocol in breast cancer patients. However, the main challenge to be further investigated still remains the emergence of therapeutic resistance mechanisms, which develop soon after the onset of therapy and need urgent attention and further elucidation. What are the recent emerging molecular resistance mechanisms in breast cancer targeted therapy and what are the best strategies to apply in order to circumvent this important obstacle? The main scope of this review is to provide a thorough update of recent developments in the field and discuss future prospects for preventing resistance mechanisms in the quest to increase overall survival of patients suffering from the disease.
Collapse
|
29
|
Wong F, Coban O, Weitsman G, Ng T. Integrating imaging, exosome and protein network rewiring information to track early tumour evolution of resistance mechanisms. CONVERGENT SCIENCE PHYSICAL ONCOLOGY 2017. [DOI: 10.1088/2057-1739/aa5cbd] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
30
|
Vani K, Sompuram SR, Schaedle AK, Balasubramanian A, Bogen SA. Analytic Response Curves of Clinical Breast Cancer IHC Tests. J Histochem Cytochem 2017; 65:273-283. [PMID: 28438091 DOI: 10.1369/0022155417694869] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
An important limitation in the field of immunohistochemistry (IHC) is the inability to correlate stain intensity with specific analyte concentrations. Clinical immunohistochemical tests are not described in terms of analytic response curves, namely, the analyte concentrations in a tissue sample at which an immunohistochemical stain (1) is first visible, (2) increases in proportion to the analyte concentration, and (3) ultimately approaches a maximum color intensity. Using a new immunostaining tool ( IHControls), we measured the analytic response curves of the major clinical immunohistochemical tests for human epidermal growth factor receptor type II (HER-2), estrogen receptor (ER), and progesterone receptor (PR). The IHControls comprise the analytes HER-2, ER, and PR at approximately log concentration intervals across the range of biological expression, from 100 to 1,000,000 molecules per test microbead. We stained IHControls of various concentrations using instruments, reagents, and protocols from three major IHC vendors. Stain intensity at each analyte concentration was measured, thereby generating an analytic response curve. We learned that for HER-2 and PR, there is significant variability in test results between clinical kits for samples with analyte concentrations of approximately 104 molecules/microbead. We propose that the characterization of immunostains is an important step toward standardization.
Collapse
Affiliation(s)
- Kodela Vani
- Medical Discovery Partners LLC, Boston, Massachusetts (KV, SRS, AKS, AB, SAB), and Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts (SAB)
| | - Seshi R Sompuram
- Medical Discovery Partners LLC, Boston, Massachusetts (KV, SRS, AKS, AB, SAB), and Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts (SAB)
| | - Anika K Schaedle
- Medical Discovery Partners LLC, Boston, Massachusetts (KV, SRS, AKS, AB, SAB), and Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts (SAB)
| | - Anuradha Balasubramanian
- Medical Discovery Partners LLC, Boston, Massachusetts (KV, SRS, AKS, AB, SAB), and Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts (SAB)
| | - Steven A Bogen
- Medical Discovery Partners LLC, Boston, Massachusetts (KV, SRS, AKS, AB, SAB), and Department of Pathology and Laboratory Medicine, Tufts Medical Center, Boston, Massachusetts (SAB)
| |
Collapse
|
31
|
Predicting and Overcoming Chemotherapeutic Resistance in Breast Cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1026:59-104. [PMID: 29282680 DOI: 10.1007/978-981-10-6020-5_4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Our understanding of breast cancer and its therapeutic approach has improved greatly due to the advancement of molecular biology in recent years. Clinically, breast cancers are characterized into three basic types based on their immunohistochemical properties. They are triple-negative breast cancer, estrogen receptor (ER) and progesterone receptor (PR)-positive-HR positive breast cancer, and human epidermal growth factor receptor 2 (HER2)-positive breast cancer. Even though these subtypes have been characterized, assessment of a breast cancer's receptor status is still widely used to determine whether or not a targeted therapy could be applied. Moreover, drug resistance is common in all breast cancer types despite the different treatment modalities applied. The development of resistance to different therapeutics is not mutually exclusive. It seems that tumor could be resistant to multiple treatment strategies, such as being both chemoresistant and monoclonal antibody resistant. However, the underlying mechanisms are complicated and need further investigation. In this chapter, we aim to provide a brief review of the different types of breast cancer and their respective treatment strategies. We also review the possible mechanisms of potential drug resistance associated with each treatment type. We believe that a better understanding of the drug resistance mechanisms can lead to a more effective and efficient therapeutic success.
Collapse
|
32
|
Weitsman G, Barber PR, Nguyen LK, Lawler K, Patel G, Woodman N, Kelleher MT, Pinder SE, Rowley M, Ellis PA, Purushotham AD, Coolen AC, Kholodenko BN, Vojnovic B, Gillett C, Ng T. HER2-HER3 dimer quantification by FLIM-FRET predicts breast cancer metastatic relapse independently of HER2 IHC status. Oncotarget 2016; 7:51012-51026. [PMID: 27618787 PMCID: PMC5239455 DOI: 10.18632/oncotarget.9963] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 05/23/2016] [Indexed: 01/08/2023] Open
Abstract
Overexpression of HER2 is an important prognostic marker, and the only predictive biomarker of response to HER2-targeted therapies in invasive breast cancer. HER2-HER3 dimer has been shown to drive proliferation and tumor progression, and targeting of this dimer with pertuzumab alongside chemotherapy and trastuzumab, has shown significant clinical utility. The purpose of this study was to accurately quantify HER2-HER3 dimerisation in formalin fixed paraffin embedded (FFPE) breast cancer tissue as a novel prognostic biomarker.FFPE tissues were obtained from patients included in the METABRIC (Molecular Taxonomy of Breast Cancer International Consortium) study. HER2-HER3 dimerisation was quantified using an improved fluorescence lifetime imaging microscopy (FLIM) histology-based analysis. Analysis of 131 tissue microarray cores demonstrated that the extent of HER2-HER3 dimer formation as measured by Förster Resonance Energy Transfer (FRET) determined through FLIM predicts the likelihood of metastatic relapse up to 10 years after surgery (hazard ratio 3.91 (1.61-9.5), p = 0.003) independently of HER2 expression, in a multivariate model. Interestingly there was no correlation between the level of HER2 protein expressed and HER2-HER3 heterodimer formation. We used a mathematical model that takes into account the complex interactions in a network of all four HER proteins to explain this counterintuitive finding.Future utility of this technique may highlight a group of patients who do not overexpress HER2 protein but are nevertheless dependent on the HER2-HER3 heterodimer as driver of proliferation. This assay could, if validated in a group of patients treated with, for instance pertuzumab, be used as a predictive biomarker to predict for response to such targeted therapies.
Collapse
Affiliation(s)
- Gregory Weitsman
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer Studies, King's College London, Guy's Medical School Campus, London, UK
| | - Paul R. Barber
- Department of Oncology, Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
- Institute for Mathematical and Molecular Biomedicine, King's College London, Guy's Medical School Campus, London, UK
| | - Lan K. Nguyen
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
- Department of Biochemistry and Molecular Biology, School of Biomedical Sciences and Biomedical Discovery Institute, Monash University, Melbourne, Australia
| | - Katherine Lawler
- Institute for Mathematical and Molecular Biomedicine, King's College London, Guy's Medical School Campus, London, UK
| | - Gargi Patel
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer Studies, King's College London, Guy's Medical School Campus, London, UK
- Sussex Cancer Centre, Brighton and Sussex University Hospitals, Royal Sussex County Hospital, Brighton, UK
| | - Natalie Woodman
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, Great Maze Pond, London, UK
- Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital King's College London School of Medicine, London, UK
| | - Muireann T. Kelleher
- Department of Medical Oncology, St George's Hospital NHS Foundation Trust, London, UK
| | - Sarah E. Pinder
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, Great Maze Pond, London, UK
- Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital King's College London School of Medicine, London, UK
| | - Mark Rowley
- Institute for Mathematical and Molecular Biomedicine, King's College London, Guy's Medical School Campus, London, UK
| | - Paul A. Ellis
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, Great Maze Pond, London, UK
| | - Anand D. Purushotham
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, Great Maze Pond, London, UK
| | - Anthonius C. Coolen
- Institute for Mathematical and Molecular Biomedicine, King's College London, Guy's Medical School Campus, London, UK
| | - Boris N. Kholodenko
- Systems Biology Ireland, University College Dublin, Belfield, Dublin, Ireland
| | - Borivoj Vojnovic
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer Studies, King's College London, Guy's Medical School Campus, London, UK
- Department of Oncology, Cancer Research UK and Medical Research Council Oxford Institute for Radiation Oncology, University of Oxford, Oxford, UK
| | - Cheryl Gillett
- Research Oncology, Division of Cancer Studies, King's College London, Guy's Hospital, Great Maze Pond, London, UK
| | - Tony Ng
- Richard Dimbleby Department of Cancer Research, Randall Division and Division of Cancer Studies, King's College London, Guy's Medical School Campus, London, UK
- Breakthrough Breast Cancer Research Unit, Department of Research Oncology, Guy's Hospital King's College London School of Medicine, London, UK
- UCL Cancer Institute, Paul O'Gorman Building, University College London, London, UK
| |
Collapse
|
33
|
Fotticchia I, Guarnieri D, Fotticchia T, Falanga AP, Vecchione R, Giancola C, Netti PA. Energetics of ligand-receptor binding affinity on endothelial cells: An in vitro model. Colloids Surf B Biointerfaces 2016; 144:250-256. [DOI: 10.1016/j.colsurfb.2016.04.018] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 03/18/2016] [Accepted: 04/07/2016] [Indexed: 01/31/2023]
|
34
|
Lédel F, Stenstedt K, Hallström M, Ragnhammar P, Edler D. HER3 expression is correlated to distally located and low-grade colon cancer. Acta Oncol 2016; 55:875-80. [PMID: 26863446 DOI: 10.3109/0284186x.2015.1131334] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
UNLABELLED Background HER3 is a member of the human epidermal growth factor receptor complex (EGFR, HER2, HER3 and HER4). It has been investigated as a prognostic biomarker in colorectal cancer but is sparingly studied in colon cancer. HER3 can affect cellular proliferation, differentiation and migration in oncogenesis through ligand binding and activation of intracellular signal pathways. Recently, we found that expression of cell surface HER3 can be detected at a high extent in primary colorectal tumors, lymph node and liver metastases and that it correlated with poor prognosis. This large, explorative, retrospective study evaluates the prognostic value of HER3 in colon cancer and the association of HER3 to tumor location. MATERIAL AND METHODS Immunohistochemical detection with a monoclonal HER3 antibody in primary colon tumors of stage II and III, from 521 patients, was performed. Results HER3 was expressed at high levels in 67% of the colon tumors. High HER3 expression was associated with distal tumor location (p < 0.0001) and low-grade tumor (p < 0.0001). In the group of patients with distal colon cancer (230/521), HER3 expression correlated to shorter disease-free survival (DFS) (p = 0.03) in the univariate analysis and in the multivariate analysis, a hazard ratio of 0.56 (95% CI 0.31-0.99) (p = 0.047) was observed. Conclusion In this explorative, retrospective study, high HER3 expression in colon cancer was associated to distal colon location and low-grade tumor. High HER3 expression was of prognostic value according to DFS in distal colon cancer in univariate and multivariate analysis. We could not find a significant value of HER3 expression with respect to overall survival (OS).
Collapse
Affiliation(s)
- Frida Lédel
- Institution of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Gastrocentrum, Department of Colorectal Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Kristina Stenstedt
- Institution of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Gastrocentrum, Department of Colorectal Surgery, Karolinska University Hospital, Stockholm, Sweden
| | - Marja Hallström
- Institution of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Peter Ragnhammar
- Institution of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - David Edler
- Institution of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden
- Gastrocentrum, Department of Colorectal Surgery, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
35
|
Ki J, Arumugam P, Song JM. TIRF high-content assay development for the evaluation of drug efficacy of chemotherapeutic agents against EGFR-/HER2-positive breast cancer cell lines. Anal Bioanal Chem 2016; 408:3233-8. [PMID: 26886741 DOI: 10.1007/s00216-016-9387-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Accepted: 02/02/2016] [Indexed: 01/06/2023]
Abstract
Elevated expression of epidermal growth factor receptor (EGFR) is reported to be associated with poor prognosis in breast cancer. EGFR subtype identification plays a crucial role in deciding the drug combination to treat the cancer patients. Conventional application of immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) produces more discordance results in EGFR subtype identification of cancer specimens. The present study is designed to develop an analytical method for simultaneous identification of cell surface biomarkers and quantitative estimation of drug efficacy in cancer specimens. For this study, we have utilized a total internal reflection fluorescence microscope (TIRFM), Qdot molecular probes and chemotherapeutic agent camptothecin (CPT)-treated breast cancer cell lines namely MCF-7, SK-BR-3 and JIMT-1. Highly sensitive detection signals with low background noise generated from the evanescent field excitation of TIRFM make it a highly suitable tool to detect the cell surface biomarkers in living cells. Moreover, single wavelength excitation of Qdot probes offers multicolour imaging with strong emission brightness. In the present study, TIRF high-content imaging system simultaneously showed the expression pattern of EGFRs and EC50 value for CPT-induced apoptosis and necrosis in MCF-7, SK-BR-3 and JIMT-1 cancer cell lines.
Collapse
Affiliation(s)
- Jieun Ki
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-ku, Seoul, 151-742, Republic of Korea
| | - Parthasarathy Arumugam
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-ku, Seoul, 151-742, Republic of Korea
| | - Joon Myong Song
- College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-ku, Seoul, 151-742, Republic of Korea.
| |
Collapse
|
36
|
Boellner S, Becker KF. Recent progress in protein profiling of clinical tissues for next-generation molecular diagnostics. Expert Rev Mol Diagn 2015. [DOI: 10.1586/14737159.2015.1070098] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|